Feature Papers in Cancer Biology and Therapeutics

A topical collection in Biomedicines (ISSN 2227-9059). This collection belongs to the section "Cancer Biology and Oncology".

Viewed by 4575

Editor


E-Mail Website
Collection Editor
1. NF-kB, Differenciation and Cancer, University Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
2. Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
Interests: interface between signal transduction and cancer with a focus on the alternative NF-kappaB signaling pathway, how it is regulated, and its contributions towards tumor development and resistance to conventional cancer therapies
Special Issues, Collections and Topics in MDPI journals

Topical Collection Information

Dear Colleagues,

The Topical Collection entitled “Feature Papers in Cancer Biology and Therapeutics” aims to collect high-quality research articles and review articles covering all the aspects of cancer biology research and innovative cancer therapies.

The topics include, but are not limited to, a fundamental understanding of biological processes underlying cancer initiation, progression, metastasis and resistance to treatment, cancer cell metabolism, tumor microenvironment, gene expression profiling, epigenetic regulation, oncogenetics, omics approaches, diagnostic and prognostic biomarkers, molecular targets, cancer drug development, clinical trials with new agents, and validation in animal models. We look forward to your submissions on the above-listed research areas of cancer biology and therapies.

Dr. Veronique Baud
Collection Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the collection website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomedicines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • molecular and cellular basis of cancer
  • metastasis
  • resistance to treatment
  • cancer metabolism
  • stress responses
  • tumor microenvironment
  • cancer genome
  • precision oncology
  • cancer related to:
    • cell death
    • cell growth
    • invasion
    • gene expression profiling
    • metabolomics
    • epigenetics
    • oncogenomics
    • biomarkers
    • molecular targets
    • cancer-targeted diagnosis
    • cancer-targeted therapeutics
    • innovative anti-cancer therapies
    • mechanism-based drug development
    • drug discovery
    • clinical trials
    • animal models

Published Papers (3 papers)

2024

Jump to: 2022

19 pages, 9251 KiB  
Article
Analysing the Combined Effects of Radiotherapy and Chemokine Receptor 5 Antagonism: Complementary Approaches to Promote T Cell Function and Migration in Oesophageal Adenocarcinoma
by Maria Davern, Cillian O’ Donovan, Noel E. Donlon, Eimear Mylod, Caoimhe Gaughan, Anshul Bhardwaj, Andrew D. Sheppard, Dara Bracken-Clarke, Christine Butler, Narayanasamy Ravi, Claire L. Donohoe, John V. Reynolds, Joanne Lysaght and Melissa J. Conroy
Biomedicines 2024, 12(4), 819; https://doi.org/10.3390/biomedicines12040819 - 08 Apr 2024
Viewed by 308
Abstract
The presence of an immunosuppressive tumour microenvironment in oesophageal adenocarcinoma (OAC) is a major contributor to poor responses. Novel treatment strategies are required to supplement current regimens and improve patient survival. This study examined the immunomodulatory effects that radiation therapy and chemokine receptor [...] Read more.
The presence of an immunosuppressive tumour microenvironment in oesophageal adenocarcinoma (OAC) is a major contributor to poor responses. Novel treatment strategies are required to supplement current regimens and improve patient survival. This study examined the immunomodulatory effects that radiation therapy and chemokine receptor antagonism impose on T cell phenotypes in OAC with a primary goal of identifying potential therapeutic targets to combine with radiation to improve anti-tumour responses. Compared with healthy controls, anti-tumour T cell function was impaired in OAC patients, demonstrated by lower IFN-γ production by CD4+ T helper cells and lower CD8+ T cell cytotoxic potential. Such diminished T cell effector functions were enhanced following treatment with clinically relevant doses of irradiation. Interestingly, CCR5+ T cells were significantly more abundant in OAC patient blood compared with healthy controls, and CCR5 surface expression by T cells was further enhanced by clinically relevant doses of irradiation. Moreover, irradiation enhanced T cell migration towards OAC patient-derived tumour-conditioned media (TCM). In vitro treatment with the CCR5 antagonist Maraviroc enhanced IFN-γ production by CD4+ T cells and increased the migration of irradiated CD8+ T cells towards irradiated TCM, suggesting its synergistic therapeutic potential in combination with irradiation. Overall, this study highlights the immunostimulatory properties of radiation in promoting anti-tumour T cell responses in OAC and increasing T cell migration towards chemotactic cues in the tumour. Importantly, the CCR5 antagonist Maraviroc holds promise to be repurposed in combination with radiotherapy to promote anti-tumour T cell responses in OAC. Full article
Show Figures

Figure 1

20 pages, 2367 KiB  
Article
An Integrated Approach Including CRISPR/Cas9-Mediated Nanopore Sequencing, Mate Pair Sequencing, and Cytogenomic Methods to Characterize Complex Structural Rearrangements in Acute Myeloid Leukemia
by Michael Phan, Maria A. Gomes, Victoria Stinnett, Laura Morsberger, Nicole L. Hoppman, Kathryn E. Pearce, Kirstin Smith, Brian Phan, Liqun Jiang and Ying S. Zou
Biomedicines 2024, 12(3), 598; https://doi.org/10.3390/biomedicines12030598 - 07 Mar 2024
Viewed by 645
Abstract
Complex structural chromosome abnormalities such as chromoanagenesis have been reported in acute myeloid leukemia (AML). They are usually not well characterized by conventional genetic methods, and the characterization of chromoanagenesis structural abnormalities from short-read sequencing still presents challenges. Here, we characterized complex structural [...] Read more.
Complex structural chromosome abnormalities such as chromoanagenesis have been reported in acute myeloid leukemia (AML). They are usually not well characterized by conventional genetic methods, and the characterization of chromoanagenesis structural abnormalities from short-read sequencing still presents challenges. Here, we characterized complex structural abnormalities involving chromosomes 2, 3, and 7 in an AML patient using an integrated approach including CRISPR/Cas9-mediated nanopore sequencing, mate pair sequencing (MPseq), and SNP microarray analysis along with cytogenetic methods. SNP microarray analysis revealed chromoanagenesis involving chromosomes 3 and 7, and a pseudotricentric chromosome 7 was revealed by cytogenetic methods. MPseq revealed 138 structural variants (SVs) as putative junctions of complex rearrangements involving chromosomes 2, 3, and 7, which led to 16 novel gene fusions and 33 truncated genes. Thirty CRISPR RNA (crRNA) sequences were designed to map 29 SVs, of which 27 (93.1%) were on-target based on CRISPR/Cas9 crRNA nanopore sequencing. In addition to simple SVs, complex SVs involving over two breakpoints were also revealed. Twenty-one SVs (77.8% of the on-target SVs) were also revealed by MPseq with shared SV breakpoints. Approximately three-quarters of breakpoints were located within genes, especially intronic regions, and one-quarter of breakpoints were intergenic. Alu and LINE repeat elements were frequent among breakpoints. Amplification of the chromosome 7 centromere was also detected by nanopore sequencing. Given the high amplification of the chromosome 7 centromere, extra chromosome 7 centromere sequences (tricentric), and more gains than losses of genomic material, chromoanasynthesis and chromothripsis may be responsible for forming this highly complex structural abnormality. We showed this combination approach’s value in characterizing complex structural abnormalities for clinical and research applications. Characterization of these complex structural chromosome abnormalities not only will help understand the molecular mechanisms responsible for the process of chromoanagenesis, but also may identify specific molecular targets and their impact on therapy and overall survival. Full article
Show Figures

Figure 1

2022

Jump to: 2024

11 pages, 1193 KiB  
Review
Harnessing the Power of Mucosal-Associated Invariant T (MAIT) Cells in Cancer Cell Therapy
by Chie Sugimoto, Hiroyoshi Fujita and Hiroshi Wakao
Biomedicines 2022, 10(12), 3160; https://doi.org/10.3390/biomedicines10123160 - 07 Dec 2022
Cited by 1 | Viewed by 2067
Abstract
Mucosal-associated invariant T (MAIT) cells, a burgeoning type of the innate-like T cells, play a crucial role in maintaining immune homeostasis, particularly in host defense. Although many studies have implied the use of MAIT cells in tumor immunity, whether MAIT cells are pro-tumor [...] Read more.
Mucosal-associated invariant T (MAIT) cells, a burgeoning type of the innate-like T cells, play a crucial role in maintaining immune homeostasis, particularly in host defense. Although many studies have implied the use of MAIT cells in tumor immunity, whether MAIT cells are pro-tumor or anti-tumor has remained elusive, as in the case for other innate-like T cells that possess dichotomous roles in tumor immunity. Although this difficulty persists where endogenous MAIT cells are the target for therapeutic intervention, the advent of induced pluripotent stem-cell-derived MAIT cells (reMAIT cells) will make it possible to harness these cells for immune cell therapy. In this review, we will discuss possible roles of MAIT cells in tumor immunity and the potential of reMAIT cells to treat tumors. Full article
Show Figures

Figure 1

Back to TopTop